Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add filters

Language
Document Type
Year range
2.
medrxiv; 2022.
Preprint in English | medRxiv | ID: ppzbmed-10.1101.2022.02.17.22271057

ABSTRACT

The SARS-CoV-2 infection is associated with increased levels of autoantibodies targeting immunological proteins such as cytokines and chemokines. Reports further indicate that COVID-19 patients may develop a wide spectrum of autoimmune diseases due to reasons not fully understood. Even so, the landscape of autoantibodies induced by SARS-CoV-2 infection remains uncharted territory. To gain more insight, we carried out a comprehensive assessment of autoantibodies known to be linked to diverse autoimmune diseases observed in COVID-19 patients, in a cohort of 248 individuals, of which 171 were COVID-19 patients (74 with mild, 65 moderate, and 32 with severe disease) and 77 were healthy controls. Dysregulated autoantibody serum levels, characterized mainly by elevated concentrations, occurred mostly in patients with moderate or severe COVID-19 infection, and was accompanied by a progressive disruption of physiologic IgG and IgA autoantibody signatures. A similar perturbation was found in patients with anosmia. Notably, autoantibody levels often accompanied anti-SARS-CoV-2 antibody concentrations, being both indicated by random forest classification as strong predictors of COVID-19 outcome, together with age. Moreover, higher levels of autoantibodies (mainly IgGs) were seen in the elderly with severe disease compared with young COVID-19 patients with severe disease. These findings suggest that the SARS-CoV-2 infection induces a broader loss of self-tolerance than previously thought, providing new ideas for therapeutic interventions.


Subject(s)
COVID-19 , Olfaction Disorders , Autoimmune Diseases
3.
ssrn; 2021.
Preprint in English | PREPRINT-SSRN | ID: ppzbmed-10.2139.ssrn.3945023

ABSTRACT

Clinical and hyperinflammatory overlap between COVID-19 and hemophagocytic lymphohistiocytosis (HLH) has been reported. However, the underlying mechanisms are unclear. Here we show that COVID-19 and HLH have an overlap of signaling pathways and gene signatures commonly dysregulated, which were defined by investigating the transcriptomes of 1253 subjects (controls, COVID-19, and HLH patients) using microarray, bulk RNA-sequencing (RNAseq), and single-cell RNAseq (scRNAseq). COVID-19 and HLH share pathways involved in cytokine and chemokine signaling as well as neutrophil-mediated immune responses that associate with COVID-19 severity. These genes are dysregulated at protein level across several COVID-19 studies and form an interconnected network with differentially expressed plasma proteins which converge to neutrophil hyperactivation in COVID-19 patients admitted to the intensive care unit. scRNAseq analysis indicated that these genes are specifically upregulated across different leukocyte populations, including lymphocyte subsets and immature neutrophils. Artificial intelligence modeling confirmed the strong association of these genes with COVID-19 severity. Thus, our work indicates putative therapeutic pathways for intervention.Funding: We acknowledge the Latin American Society of Immunodeficiencies (LASID) for providing the research funding of LFS (LASID Fellowship award 2020), and the São Paulo Research Foundation (FAPESP grants 2018/18886-9, 2020/01688-0, and 2020/07069-0 to OCM) for financial support. Computational analysis was supported by FAPESP and partially by the grants from Ontario Research Fund (#34876), Natural Sciences Research Council (NSERC #203475), Canada Foundation for Innovation (CFI #29272, #225404, #33536), and IBM granted to IJ, the National Institutes of Health (NHLBI) through award HL130704 granted to AJ, as well as the NIH P4 GM108538 granted to KAO and JJC. This study was financed in part by the coordination for the improvement of higher education personnel – Brazil (CAPES) – finance code 001.Declaration of Interests: The authors have declared that no conflict of interest exists.


Subject(s)
Smear Layer , Lymphohistiocytosis, Hemophagocytic , Immunologic Deficiency Syndromes , COVID-19
4.
medrxiv; 2020.
Preprint in English | medRxiv | ID: ppzbmed-10.1101.2020.11.12.20230417

ABSTRACT

The coronavirus disease 2019 (COVID-19) fatality rate varies in different patient groups. However, the underlying mechanisms that explain this variation are poorly understood. Here, we reanalyzed and integrated public RNAseq datasets of nasopharyngeal swabs and peripheral blood leukocytes from patients with SARS-CoV-2, comparing transcription patterns according to sex, age, and viral load. We found that female and young patients infected by SARS-CoV-2 exhibited a similar transcriptomic pattern with a larger number of total (up- and downregulated) differentially expressed genes (DEGs) compared to males and elderly patients. The transcriptional analysis showed a sex-specific profile with a higher transcriptional modulation of immune response-associated genes in female and young subjects against SARS-CoV-2. The functional clustering was characterized by a highly correlated interferome network of cytokine/chemokine- and neutrophil-associated genes that were enriched both in nasopharyngeal cells and peripheral blood of COVID-19 patients. Females exhibited reduced transcriptional levels of key pro-inflammatory/neutrophil-related genes such as CXCL8 receptors (CXCR1/CXCR2), IL-1{beta}, S100A9, ITGAM, and DBNL compared to males, which correlate with a protective gene expression profile against inflammatory damage. Our data indicate specific immune-regulatory pathways associated with sex and age of patients infected with SARS-CoV-2. These results point out therapeutic targets to reduce morbidity and mortality of COVID-19.


Subject(s)
COVID-19
SELECTION OF CITATIONS
SEARCH DETAIL